Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Biology (Basel) ; 13(3)2024 Mar 05.
Article En | MEDLINE | ID: mdl-38534438

Transient receptor potential vanilloid-6 (TRPV6) is a cation channel belonging to the TRP superfamily, specifically the vanilloid subfamily, and is the sixth member of this subfamily. Its presence in the body is primarily limited to the skin, ovaries, kidney, testes, and digestive tract epithelium. The body maintains calcium homeostasis using the TRPV6 channel, which has a greater calcium selectivity than the other TRP channels. Several pieces of evidence suggest that it is upregulated in the advanced stages of thyroid, ovarian, breast, colon, and prostate cancers. The function of TRPV6 in regulating calcium signaling in cancer will be covered in this review, along with its potential applications as a cancer treatment target.

2.
Biomed Pharmacother ; 173: 116372, 2024 Apr.
Article En | MEDLINE | ID: mdl-38432129

An increasing number of studies have shown that FAM83A, a member of the family with sequence similarity 83 (FAM83), which consists of eight members, is a key tumor therapeutic target involved in multiple signaling pathways. It has been reported that FAM83A plays essential roles in the regulation of Wnt/ß-catenin, EGFR, MAPK, EMT, and other signaling pathways and physiological processes in models of pancreatic cancer, lung cancer, breast cancer, and other malignant tumors. Moreover, the expression of FAM83A could be significantly affected by multiple noncoding RNAs that are dysregulated in malignant tumors, the dysregulation of which is essential for the malignant process. Among these noncoding RNAs, the most noteworthy is the antisense long noncoding (Lnc) RNA of FAM83A itself (FAM83A-AS1), indicating an outstanding synergistic carcinogenic effect between FAM83A and FAM83A-AS1. In the present study, the specific mechanisms by which FAM83A and FAM83A-AS1 cofunction in the Wnt/ß-catenin and EGFR signaling pathways were reviewed in detail, which will guide subsequent research. We also described the applications of FAM83A and FAM83A-AS1 in tumor therapy and provided a certain theoretical basis for subsequent drug target development and combination therapy strategies.


Lung Neoplasms , RNA, Long Noncoding , Humans , beta Catenin/genetics , beta Catenin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Movement/genetics , Lung Neoplasms/pathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , RNA, Long Noncoding/genetics , Wnt Signaling Pathway/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Neoplasm Proteins/metabolism
3.
Adv Biol (Weinh) ; 8(5): e2300117, 2024 May.
Article En | MEDLINE | ID: mdl-38379270

The incidence of Hepatocellular carcinoma (HCC) and HCC-related deaths have remarkably increased over the recent decades. It has been reported that ß-catenin activation can be frequently observed in HCC cases. This study identified the integrin-linked kinase-associated phosphatase (ILKAP) as a novel ß-catenin-interacting protein. ILKAP is localized both in the nucleus and cytoplasm and regulates the WNT pathway in different ways. First, it is demonstrated that ILKAP activates the WNT pathway in HCC cells by increasing the protein level of ß-catenin and other proteins associated with the WNT signaling, such as c-Myc and CyclinD1. Next, it is shown that ILKAP promotes the metastasis of HCC both in vitro and in vivo in a zebrafish xenograft model. It is also found that ILKAP dephosphorylates the GSK3ß and CK1, contributing to the reduced ubiquitination of ß-catenin. Furthermore, it is identified that ILKAP functions by mediating binding between TCF4 and ß-catenin to enhance expression of WNT target genes. Taken together, the study demonstrates a critical function of ILKAP in metastasis of HCC, since ILKAP is crucial for the activation of the WNT pathway via stabilization of ß-catenin and increased binding between TCF4 and ß-catenin.


Carcinoma, Hepatocellular , Liver Neoplasms , Phosphoprotein Phosphatases , Wnt Signaling Pathway , beta Catenin , Animals , Humans , beta Catenin/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Neoplasm Metastasis , Transcription Factor 4/metabolism , Transcription Factor 4/genetics , Wnt Signaling Pathway/physiology , Zebrafish , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism
4.
Cell Death Differ ; 30(10): 2231-2248, 2023 Oct.
Article En | MEDLINE | ID: mdl-37684417

Autophagy serves as a pro-survival mechanism for a cell or a whole organism to cope with nutrient stress. Our understanding of the molecular regulation of this fusion event remains incomplete. Here, we identified RUNDC1 as a novel ATG14-interacting protein, which is highly conserved across vertebrates, including zebrafish and humans. By gain and loss of function studies, we demonstrate that RUNDC1 negatively modulates autophagy by blocking fusion between autophagosomes and lysosomes via inhibiting the assembly of the STX17-SNAP29-VAMP8 complex both in human cells and the zebrafish model. Moreover, RUNDC1 clasps the ATG14-STX17-SNAP29 complex via stimulating ATG14 homo-oligomerization to inhibit ATG14 dissociation. This also prevents VAMP8 from binding to STX17-SNAP29. We further identified that phosphorylation of RUNDC1 Ser379 is crucial to inhibit the assembly of the STX17-SNAP29-VAMP8 complex via promoting ATG14 homo-oligomerization. In line with our findings, RunDC1 is crucial for zebrafish in their response to nutrient-deficient conditions. Taken together, our findings demonstrate that RUNDC1 is a negative regulator of autophagy that restricts autophagosome fusion with lysosomes by clasping the ATG14-STX17-SNAP29 complex to hinder VAMP8 binding.

5.
Biochim Biophys Acta Mol Cell Res ; 1870(7): 119537, 2023 10.
Article En | MEDLINE | ID: mdl-37463638

Macroautophagy is a health-modifying process of engulfing misfolded or aggregated proteins or damaged organelles, coating these proteins or organelles into vesicles, fusion of vesicles with lysosomes to form autophagic lysosomes, and degradation of the encapsulated contents. It is also a self-rescue strategy in response to harsh environments and plays an essential role in cancer cells. AMP-activated protein kinase (AMPK) is the central pathway that regulates autophagy initiation and autophagosome formation by phosphorylating targets such as mTORC1 and unc-51 like activating kinase 1 (ULK1). AMPK is an evolutionarily conserved serine/threonine protein kinase that acts as an energy sensor in cells and regulates various metabolic processes, including those involved in cancer. The regulatory network of AMPK is complicated and can be regulated by multiple upstream factors, such as LKB1, AKT, PPAR, SIRT1, or noncoding RNAs. Currently, AMPK is being investigated as a novel target for anticancer therapies based on its role in macroautophagy regulation. Herein, we review the effects of AMPK-dependent autophagy on tumor cell survival and treatment strategies targeting AMPK.


AMP-Activated Protein Kinases , Neoplasms , AMP-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Autophagy , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasms/genetics
6.
Oncogene ; 42(32): 2439-2455, 2023 08.
Article En | MEDLINE | ID: mdl-37400529

The Wnt/ß-catenin signaling is usually abnormally activated in hepatocellular carcinoma (HCC), and pituitary tumor-transforming gene 1 (PTTG1) has been found to be highly expressed in HCC. However, the specific mechanism of PTTG1 pathogenesis remains poorly understood. Here, we found that PTTG1 is a bona fide ß-catenin binding protein. PTTG1 positively regulates Wnt/ß-catenin signaling by inhibiting the destruction complex assembly, promoting ß-catenin stabilization and subsequent nuclear localization. Moreover, the subcellular distribution of PTTG1 was regulated by its phosphorylation status. Among them, PP2A induced PTTG1 dephosphorylation at Ser165/171 residues and prevented PTTG1 translocation into the nucleus, but these effects were effectively reversed by PP2A inhibitor okadaic acid (OA). Interestingly, we found that PTTG1 decreased Ser9 phosphorylation-inactivation of GSK3ß by competitively binding to PP2A with GSK3ß, indirectly leading to cytoplasmic ß-catenin stabilization. Finally, PTTG1 was highly expressed in HCC and associated with poor patient prognosis. PTTG1 could promote the proliferative and metastasis of HCC cells. Overall, our results indicated that PTTG1 plays a crucial role in stabilizing ß-catenin and facilitating its nuclear accumulation, leading to aberrant activation of Wnt/ß-catenin signaling and providing a feasible therapeutic target for human HCC.


Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Cell Line , Wnt Signaling Pathway/genetics , Cell Line, Tumor
7.
Cells ; 12(8)2023 04 11.
Article En | MEDLINE | ID: mdl-37190041

Autophagy is a highly conserved recycling process of eukaryotic cells that degrades protein aggregates or damaged organelles with the participation of autophagy-related proteins. Membrane bending is a key step in autophagosome membrane formation and nucleation. A variety of autophagy-related proteins (ATGs) are needed to sense and generate membrane curvature, which then complete the membrane remodeling process. The Atg1 complex, Atg2-Atg18 complex, Vps34 complex, Atg12-Atg5 conjugation system, Atg8-phosphatidylethanolamine conjugation system, and transmembrane protein Atg9 promote the production of autophagosomal membranes directly or indirectly through their specific structures to alter membrane curvature. There are three common mechanisms to explain the change in membrane curvature. For example, the BAR domain of Bif-1 senses and tethers Atg9 vesicles to change the membrane curvature of the isolation membrane (IM), and the Atg9 vesicles are reported as a source of the IM in the autophagy process. The amphiphilic helix of Bif-1 inserts directly into the phospholipid bilayer, causing membrane asymmetry, and thus changing the membrane curvature of the IM. Atg2 forms a pathway for lipid transport from the endoplasmic reticulum to the IM, and this pathway also contributes to the formation of the IM. In this review, we introduce the phenomena and causes of membrane curvature changes in the process of macroautophagy, and the mechanisms of ATGs in membrane curvature and autophagosome membrane formation.


Autophagosomes , Autophagy-Related Proteins , Autophagy , Cell Membrane , Proteolysis , Cell Membrane/chemistry , Cell Membrane/metabolism , Protein Aggregates , Autophagosomes/chemistry , Autophagosomes/metabolism , Autophagy-Related Proteins/chemistry , Autophagy-Related Proteins/metabolism , Protein Domains , Lipid Bilayers , Humans
8.
J Neurophysiol ; 129(4): 833-842, 2023 04 01.
Article En | MEDLINE | ID: mdl-36883767

Given the increasing trend of cannabis use for recreational and therapeutic purposes, a comprehensive examination of cannabis effects is warranted. The principal psychoactive constituent of cannabis, Δ-9-tetrahydrocannabinol (THC), is a potent disrupter of neurodevelopment. Nevertheless, the impact of acute exposure to THC on developing motor systems is not well-investigated. In this study, using a neurophysiological whole cell patch clamp approach we demonstrated that a 30-min exposure to THC can alter spontaneous synaptic activities at neuromuscular junctions of 5-day post-fertilized zebrafish. An increased frequency of synaptic activity and altered decay kinetic properties were documented in the THC-treated larvae. Locomotive behaviors, including swimming activity rate and C-start escape response to sound were also affected by THC. Although the THC-treated larvae displayed hyperactivity of their basal swimming levels, their escape response rate to sound stimuli was reduced. These findings suggest that acute exposure to THC can disrupt neuromuscular transmission and locomotor-driven responses in developing zebrafish.NEW & NOTEWORTHY Acute exposure to THC alters motor neuron-muscle communication and motor behaviors in developing zebrafish. Our neurophysiology data indicated that the properties of spontaneous synaptic activity at neuromuscular junctions, such as decay component of acetylcholine receptors and frequency of synaptic events, were affected by a 30-min exposure to THC. Hyperactivity and reduced responsiveness to the sound stimulus were also observed in the THC-treated larvae. Exposure to THC during early developing stages may induce motor dysfunction.


Dronabinol , Zebrafish , Animals , Dronabinol/pharmacology , Cannabinoid Receptor Agonists , Synaptic Transmission , Motor Neurons
9.
Signal Transduct Target Ther ; 8(1): 66, 2023 02 17.
Article En | MEDLINE | ID: mdl-36797256

Abnormal activation of Wnt/ß-catenin-mediated transcription is closely associated with the malignancy of pancreatic cancer. Family with sequence similarity 83 member A (FAM83A) was shown recently to have oncogenic effects in a variety of cancer types, but the biological roles and molecular mechanisms of FAM83A in pancreatic cancer need further investigation. Here, we newly discovered that FAM83A binds directly to ß-catenin and inhibits the assembly of the cytoplasmic destruction complex thus inhibiting the subsequent phosphorylation and degradation. FAM83A is mainly phosphorylated by the SRC non-receptor kinase family member BLK (B-lymphoid tyrosine kinase) at tyrosine 138 residue within the DUF1669 domain that mediates the FAM83A-ß-catenin interaction. Moreover, FAM83A tyrosine 138 phosphorylation enhances oncogenic Wnt/ß-catenin-mediated transcription through promoting ß-catenin-TCF4 interaction and showed an elevated nucleus translocation, which inhibits the recruitment of histone deacetylases by TCF4. We also showed that FAM83A is a direct downstream target of Wnt/ß-catenin signaling and correlates with the levels of Wnt target genes in human clinical pancreatic cancer tissues. Notably, the inhibitory peptides that target the FAM83A-ß-catenin interaction significantly suppressed pancreatic cancer growth and metastasis in vitro and in vivo. Our results revealed that blocking the FAM83A cascade signaling defines a therapeutic target in human pancreatic cancer.


Neoplasm Proteins , Pancreatic Neoplasms , beta Catenin , src-Family Kinases , Humans , beta Catenin/genetics , beta Catenin/metabolism , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/genetics , Phosphorylation/genetics , Tyrosine/metabolism , Wnt Signaling Pathway/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism , Pancreatic Neoplasms
10.
Sci Rep ; 12(1): 20251, 2022 11 24.
Article En | MEDLINE | ID: mdl-36424484

Of the three primary cannabinoids in cannabis: Δ9-Tetrahydrocannabinol (Δ9-THC), cannabidiol (CBD) and cannabinol (CBN), very little is known about the actions of CBN, the primary oxidative metabolite of THC. Our goal was to determine if CBN exposure during gastrulation alters embryonic development, and if so, does it act via the canonical cannabinoid receptors. Zebrafish embryos were exposed to CBN during gastrulation and exhibited dose-dependent malformations, increased mortality, decreased locomotion and a reduction in motor neuron branching. Moreover, larva showed a significant reduction in the response to sound stimuli. CBN exposure altered the development of hair cells associated with otic vesicles and the lateral line. Pharmacological block of Cb2rs with AM 630 or JTE 907 prevented many of the CBN-induced developmental defects, while block of Cb1rs with AM 251 or CP 945598 had little or no effect. Altogether we show that embryonic exposure to CBN results in alterations in embryonic growth, neuronal and hair cell development, physiology and behavior via Cb2r-mediated mechanisms.


Cannabinoids , Cannabinol , Animals , Cannabinol/metabolism , Zebrafish/metabolism , Cannabinoids/pharmacology , Cannabinoids/metabolism , Dronabinol/pharmacology , Dronabinol/metabolism , Receptors, Cannabinoid
11.
Dev Neurosci ; 44(6): 518-531, 2022.
Article En | MEDLINE | ID: mdl-35728564

Fast excitatory synaptic transmission in the CNS is mediated by the neurotransmitter glutamate, binding to and activating AMPA receptors (AMPARs). AMPARs are known to interact with auxiliary proteins that modulate their behavior. One such family of proteins is the transmembrane AMPAR-related proteins, known as TARPs. Little is known about the role of TARPs during development or about their function in nonmammalian organisms. Here, we report on the presence of TARP γ-4 in developing zebrafish. We find that zebrafish express 2 forms of TARP γ-4: γ-4a and γ-4b as early as 12 h post-fertilization. Sequence analysis shows that both γ-4a and γ-4b shows great level of variation particularly in the intracellular C-terminal domain compared to rat, mouse, and human γ-4. RT-qPCR showed a gradual increase in the expression of γ-4a throughout the first 5 days of development, whereas γ-4b levels were constant until day 5 when levels increased significantly. Knockdown of TARP γ-4a and γ-4b via either splice-blocking morpholinos or translation-blocking morpholinos resulted in embryos that exhibited deficits in C-start escape responses, showing reduced C-bend angles. Morphant larvae displayed reduced bouts of swimming. Whole-cell patch-clamp recordings of AMPAR-mediated currents from Mauthner cells showed a reduction in the frequency of mEPCs but no change in amplitude or kinetics. Together, these results suggest that γ-4a and γ-4b are required for proper neuronal development.


Membrane Proteins , Receptors, AMPA , Synaptic Transmission , Zebrafish Proteins , Zebrafish , Animals , Membrane Proteins/metabolism , Morpholinos , Nuclear Proteins/metabolism , Receptors, AMPA/chemistry , Receptors, AMPA/metabolism , Synaptic Transmission/physiology , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
12.
Dev Neurosci ; 44(3): 142-152, 2022.
Article En | MEDLINE | ID: mdl-35168237

The endocannabinoid system (ECS) is widely studied due to its interactions with cannabis and its role in modulating physiological responses. While most research has focused on the effects of cannabis on adult ECSs, recent studies have begun to investigate the role of the ECS in developing organisms. However, little is known about the spatial or temporal expression of these receptors during early development. This study combines reverse transcriptase-polymerase chain reaction (RT-PCR) with in situ hybridizations to compile a timeline of the developmental expression of six key cannabinoid receptors; cb1, cb2, trpv1, trpa1a, trpa1b, and gpr55 in zebrafish embryos, starting from as early as 6-h postfertilization (hpf) until 3 days pf (dpf). This time frame is roughly equivalent to 2-10 weeks in human embryonic development. All six genes were confirmed to be expressed within this time range and share similarities with human and rodent expression. Cb1 expression was first detected between 12 and 24 hpf in the retina and CNS, and its expression increased thereafter and was more evident in the olfactory bulb, tegmentum, hypothalamus, and gut. Cb2 expression was relatively high at the 6 and 24 hpf timepoints, as determined by RT-PCR but was undetectable at other times. Trpv1 was first detected at 1 dpf in the trigeminal (Tg) ganglia, Rohon-Beard neurons, and lateral line, and its expression increased in the first 3 dpf. Expression of trpa1a was first detected as late as 3 dpf in vagal (V) neurons, whereas trpa1b was first detected at 1 dpf associated with Tg, glossopharyngeal, and V ganglia. Expression of gpr55 was diffuse and widespread throughout the brain and head region but was undetectable elsewhere in the embryo. Thus, receptor expression was found to be enriched in the central nervous system and within sensory neurons. This work aims to serve as a foundation for further investigation on the role of cannabinoid and cannabinoid-interacting receptors in early embryonic development.


Cannabinoids , Zebrafish , Animals , Endocannabinoids/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , Zebrafish/genetics , Zebrafish/metabolism
13.
Cell Insight ; 1(4): 100045, 2022 Aug.
Article En | MEDLINE | ID: mdl-37192859

Epidermal growth factor receptor (EGFR) plays critical roles in cell proliferation and tumorigenesis. Autophagy has emerged as a potential mechanism involved in the acquired resistance to anti-EGFR treatments, however, the molecular mechanisms has not been fully addressed. In this study, we identified EGFR interacts with STYK1, a positive autophagy regulator, in EGFR kinase activity dependent manner. We found that EGFR phosphorylates STYK1 at Y356 site and STYK1 inhibits activated EGFR mediated Beclin1 tyrosine phosphorylation and interaction between Bcl2 and Beclin1, thus enhances PtdIns3K-C1 complex assembly and autophagy initiation. We also demonstrated that STYK1 depletion increased the sensitivity of NSCLC cells to EGFR-TKIs in vitro and in vivo. Moreover, EGFR-TKIs induced activation of AMPK phosphorylates STYK1 at S304 site. STYK1 S304 collaborated with Y356 phosphorylation to enhance the EGFR-STYK1 interaction and reverse the inhibitory effects of EGFR to autophagy flux. Collectively, these data revealed new roles and cross-talk between STYK1 and EGFR in autophagy regulation and EGFR-TKIs sensitivity in NSCLC.

14.
Front Pharmacol ; 13: 1053602, 2022.
Article En | MEDLINE | ID: mdl-36733375

Systemic lupus erythematosus (SLE) is a common multisystem, multiorgan heterozygous autoimmune disease. The main pathological features of the disease are autoantibody production and immune complex deposition. Autophagy is an important mechanism to maintain cell homeostasis. Autophagy functional abnormalities lead to the accumulation of apoptosis and induce the autoantibodies that result in immune disorders. Therefore, improving autophagy may alleviate the development of SLE. For SLE, glucocorticoids or immunosuppressive agents are commonly used in clinical treatment, but long-term use of these drugs causes serious side effects in humans. Immunosuppressive agents are expensive. Traditional Chinese medicines (TCMs) are widely used for immune diseases due to their low toxicity and few side effects. Many recent studies found that TCM and its active ingredients affected the pathological development of SLE by regulating autophagy. This article explains how autophagy interferes with immune system homeostasis and participates in the occurrence and development of SLE. It also summarizes several studies on TCM-regulated autophagy intervention in SLE to generate new ideas for basic research, the development of novel medications, and the clinical treatment of SLE.

15.
Autophagy ; 17(10): 3175-3195, 2021 10.
Article En | MEDLINE | ID: mdl-32972302

Pancreatic cancer is one of the most aggressive tumors associated with a poor clinical prognosis, weakly effective therapeutic options. Therefore, there is a strong impetus to discover new therapeutic targets in pancreatic cancer. In the present study, we first demonstrated that TSPAN1 is upregulated in pancreatic cancer and that TSPAN1 depletion decreases pancreatic cancer cell proliferation in vitro and in vivo. TSPAN1 expression was correlated with poor overall survival of pancreatic cancer patients. Moreover, we demonstrated that TSPAN1 is a novel positive regulator of macroautophagy/autophagy characterized by decreased LC3-II and SQSTM1/p62 expressions, inhibited puncta formation of GFP-LC3 and autophagic vacuoles. We also demonstrated that tspan1 mutation impaired autophagy in the zebrafish model. Furthermore, we showed that TSPAN1 promoted autophagy maturation via direct binding to LC3 by two conserved LIR motifs. Mutations in the LIR motifs of TSPAN1 resulted in a loss of the ability to induce autophagy and promote pancreatic cancer proliferation. Second, we discovered two conservative TCF/LEF binding elements present in the promoter region of the TSPAN1 gene, which was further verified through luciferase activity and ChIP assays. Furthermore, TSPAN1 was upregulated by FAM83A through the canonical WNT-CTNNB1 signaling pathway. We further demonstrated that both TSPAN1 and FAM83A are both direct targets of MIR454 (microRNA 454). Additionally, we revealed the role of MIR454-FAM83A-TSPAN1 in the proliferation of pancreatic cancer cells in vitro and in vivo. Our findings suggest that components of the MIR454-FAM83A-TSPAN1 axis may be valuable prognosis markers or therapeutic targets for pancreatic cancer.Abbreviations: AMPK: adenosine 5'-monophosphate (AMP)-activated protein kinase; APC: APC regulator of WNT signaling pathway; ATG: autophagy related; AXIN2: axin 2; BECN1: beclin 1; CCND1: cyclin D1; CSNK1A1/CK1α: casein kinase 1 alpha 1; CTNNB1/ß-catenin: catenin beta 1; DAPI: 4'6-diamino-2-phenylindole; EBSS: Earle's balanced salt solution; EdU: 5-ethynyl-20-deoxyuridine; FAM83A: family with sequence similarity 83 member A; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GSEA: gene set enrichment analysis; GSK3B: glycogen synthase kinase 3 beta; IHC: immunohistochemical; LAMP1: lysosomal associated membrane protein 1; LIR: LC3-interacting region; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; MIR454: microRNA 454; miRNA: microRNA; MKI67: antigen identified by monoclonal antibody Ki 67; MTOR: mechanistic target of rapamycin kinase; MTT: 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide; MYC: MYC proto-oncogene, bHLH transcription factor; OS: overall survival; PDAC: pancreatic ductal adenocarcinoma; RAB7A: RAB7A, member RAS oncogene family; shRNA: short hairpin RNA; SQSTM1: sequestosome 1; TBE: TCF/LEF binding element; TCGA: The Cancer Genome Atlas; TCF/LEF: transcription factor/lymphoid enhancer binding factor; TCF4: transcription factor 4; TSPAN1: tetraspanin 1; TUNEL: terminal deoxynucleotidyl transferase mediated dUTP nick end labeling; UTR: untranslated region; WT: wild type.


Autophagy , MicroRNAs , Neoplasm Proteins , Pancreatic Neoplasms , Wnt Signaling Pathway , Animals , Humans , MicroRNAs/genetics , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Tetraspanins/genetics , Zebrafish , beta Catenin/metabolism
16.
J Cell Physiol ; 236(4): 2934-2949, 2021 04.
Article En | MEDLINE | ID: mdl-33037615

Transient receptor potential melastatin member 8 (TRPM8), a Ca2+ -permeable nonselective cation channel activated by cold and cooling agents, mediates allodynia. Dysfunction or abnormal expression of TRPM8 has been found in several human cancers. The role of ubiquitination in the regulation of TRPM8 function remains poorly understood. Here, we identified the ubiquitin (Ub)-ligase E3, tripartite motif-containing 4 (TRIM4), as a novel interaction partner of TRPM8 and confirmed that the TRIM4-TRPM8 interaction was mediated through the SPRY domain of TRIM4. Patch-clamp assays showed that TRIM4 negatively regulates TRPM8-mediated currents in HEK293 cells. Moreover, TRIM4 reduced the expression of TRPM8 on the cell surface by promoting the K63-linked ubiquitination of TRPM8. Further analyses revealed that the TRPM8 N-terminal lysine residue at 423 was the major ubiquitination site that mediates its functional regulation by TRIM4. A Ub-activating enzyme E1, Ub-like modifier-activating enzyme 1 (UBA1), was also found to interact with TRPM8, thereby regulating its channel function and ubiquitination. In addition, knockdown of UBA1 impaired the regulation of TRPM8 ubiquitination and function by TRIM4. Thus, this study demonstrates that TRIM4 downregulates TRPM8 via K423-mediated TRPM8 ubiquitination and requires UBA1 to regulate TRPM8.


Lysine/metabolism , TRPM Cation Channels/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitination , Amino Acid Sequence , Animals , HEK293 Cells , Humans , MCF-7 Cells , Protein Binding , Protein Domains , Rats , Sequence Deletion , Tripartite Motif Proteins/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/metabolism
17.
Front Oncol ; 10: 573127, 2020.
Article En | MEDLINE | ID: mdl-33344232

The calcium-permeable cation channel TRPM8 (transient receptor potential melastatin 8) is a member of the TRP superfamily of cation channels that is upregulated in various types of cancer with high levels of autophagy, including prostate, pancreatic, breast, lung, and colon cancers. Autophagy is closely regulated by AMP-activated protein kinase (AMPK) and plays an important role in tumor growth by generating nutrients through degradation of intracellular structures. Additionally, AMPK activity is regulated by intracellular Ca2+ concentration. Considering that TRPM8 is a non-selective Ca2+-permeable cation channel and plays a key role in calcium homoeostasis, we hypothesized that TRPM8 may control AMPK activity thus modulating cellular autophagy to regulate the proliferation and migration of breast cancer cells. In this study, overexpression of TRPM8 enhanced the level of basal autophagy, whereas TRPM8 knockdown reduced the level of basal autophagy in several types of mammalian cancer cells. Moreover, the activity of the TRPM8 channel modulated the level of basal autophagy. The mechanism of regulation of autophagy by TRPM8 involves autophagy-associated signaling pathways for activation of AMPK and ULK1 and phagophore formation. Impaired AMPK abolished TRPM8-dependent regulation of autophagy. TRPM8 interacts with AMPK in a protein complex, and cytoplasmic C-terminus of TRPM8 mediates the TRPM8-AMPK interaction. Finally, basal autophagy mediates the regulatory effects of TRPM8 on the proliferation and migration of breast cancer cells. Thus, this study identifies TRPM8 as a novel regulator of basal autophagy in cancer cells acting by interacting with AMPK, which in turn activates AMPK to activate ULK1 in a coordinated cascade of TRPM8-mediated breast cancer progression.

18.
Mol Cancer ; 19(1): 118, 2020 07 29.
Article En | MEDLINE | ID: mdl-32727463

BACKGROUND: Pancreatic cancer is one of the most lethal malignancies and has an extremely poor diagnosis and prognosis. The development of resistance to gemcitabine is still a major challenge. The long noncoding RNA PVT1 was reported to be involved in carcinogenesis and chemoresistance; however, the mechanism by which PVT1 regulates the sensitivity of pancreatic cancer to gemcitabine remains poorly understood. METHODS: The viability of pancreatic cancer cells was assessed by MTT assay in vitro and xenograft tumor formation assay in vivo. The expression levels of PVT1 and miR-619-5p were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Western blotting analysis and qRT-PCR were performed to assess the protein and mRNA levels of Pygo2 and ATG14, respectively. Autophagy was explored via autophagic flux detection under confocal microscopy and autophagic vacuole investigation under transmission electron microscopy (TEM). The functional role and mechanism of PVT1 were further investigated by gain- and loss-of-function assays in vitro. RESULTS: In the present study, we demonstrated that PVT1 was up-regulated in gemcitabine-resistant pancreatic cancer cell lines. Gain- and loss-of-function assays revealed that PVT1 impaired sensitivity to gemcitabine in vitro and in vivo. We further found that PVT1 up-regulated the expression of both Pygo2 and ATG14 and thus regulated Wnt/ß-catenin signaling and autophagic activity to overcome gemcitabine resistance through sponging miR-619-5p. Moreover, we discovered three TCF/LEF binding elements (TBEs) in the promoter region of PVT1, and activation of Wnt/ß-catenin signaling mediated by the up-regulation of Pygo2 increased PVT1 expression by direct binding to the TBE region. Furthermore, PVT1 was discovered to interact with ATG14, thus promoting assembly of the autophagy specific complex I (PtdIns3K-C1) and ATG14-dependent class III PtdIns3K activity. CONCLUSIONS: These data indicate that PVT1 plays a critical role in the sensitivity of pancreatic cancer to gemcitabine and highlight its potential as a valuable target for pancreatic cancer therapy.


Adaptor Proteins, Vesicular Transport/genetics , Autophagy-Related Proteins/genetics , Autophagy/genetics , Drug Resistance, Neoplasm/genetics , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Wnt Signaling Pathway , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Humans , Mice , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Binding , RNA Interference , Xenograft Model Antitumor Assays , Gemcitabine
19.
J Vis Exp ; (79)2013 Sep 10.
Article En | MEDLINE | ID: mdl-24056693

Mauthner cells (M-cells) are large reticulospinal neurons located in the hindbrain of teleost fish. They are key neurons involved in a characteristic behavior known as the C-start or escape response that occurs when the organism perceives a threat. The M-cell has been extensively studied in adult goldfish where it has been shown to receive a wide range of excitatory, inhibitory and neuromodulatory signals(1). We have been examining M-cell activity in embryonic zebrafish in order to study aspects of synaptic development in a vertebrate preparation. In the late 1990s Ali and colleagues developed a preparation for patch clamp recording from M-cells in zebrafish embryos, in which the CNS was largely intact(2,3,4). The objective at that time was to record synaptic activity from hindbrain neurons, spinal cord neurons and trunk skeletal muscle while maintaining functional synaptic connections within an intact brain-spinal cord preparation. This preparation is still used in our laboratory today. To examine the mechanisms underlying developmental synaptic plasticity, we record excitatory (AMPA and NMDA-mediated)(5,6) and inhibitory (GABA and glycine) synaptic currents from developing M-cells. Importantly, this unique preparation allows us to return to the same cell (M-cell) from preparation to preparation to carefully examine synaptic plasticity and neuro-development in an embryonic organism. The benefits provided by this preparation include 1) intact, functional synaptic connections onto the M-cell, 2) relatively inexpensive preparations, 3) a large supply of readily available embryos 4) the ability to return to the same cell type (i.e. M-cell) in every preparation, so that synaptic development at the level of an individual cell can be examined from fish to fish, and 5) imaging of whole preparations due to the transparent nature of the embryos.


Neurons/physiology , Patch-Clamp Techniques/methods , Rhombencephalon/physiology , Spinal Cord/physiology , Action Potentials/physiology , Animals , Embryo, Nonmammalian , Neurons/cytology , Receptors, AMPA/physiology , Receptors, GABA-A/physiology , Receptors, Glycine/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Rhombencephalon/cytology , Synapses/physiology , Zebrafish
20.
J Neurobiol ; 62(4): 425-38, 2005 Mar.
Article En | MEDLINE | ID: mdl-15547934

Protein kinase C (PKC) is a family of enzymes involved in a wide range of biological functions. We investigated the expression of PKC-positive cells in zebrafish embryos and larvae within the first week of development to determine the developmental profile of PKC-containing cells. Our other goal was to determine if PKC alpha was associated with Rohon-Beard neurons during the first 5 days of development, when they are reported to undergo apoptosis. First, we confirmed the specificity of the antibodies by Western blotting zebrafish brain homogenates with anti-PKC and anti-PKC alpha, and detected single protein bands of approximately 78-82 kDa in size. Immunohistochemistry showed that several types of neurons were labeled, including neurons in the trigeminal ganglia, the dorsal spinal cord, and the dorsal root ganglia. Double-labeling with anti-PKC alpha and both anti-Islet-1 and zn12 confirmed the identity of the PKC-positive cells in the brain as trigeminal neurons, and in the spinal cord as Rohon-Beard cells. Some Rohon-Beard cells were labeled with anti-PKC alpha up to 7 days post fertilization (dpf). We performed TUNEL labeling and found no correlation between TUNEL-labeled and PKC alpha-labeled Rohon-Beard cells, suggesting that PKC alpha is not involved in Rohon-Beard apoptosis. Only approximately 40% of the approximately 130 Rohon-Beard cells at 24 h postfertilization (hpf) were positively labeled for PKC. Mauthner cells were labeled by anti-PKC, but not anti-PKC alpha, suggesting that the major form of PKC within these cells was not PKC alpha.


Gene Expression Regulation, Developmental/physiology , Protein Kinase C/biosynthesis , Zebrafish/embryology , Zebrafish/metabolism , Animals , Immunohistochemistry , Larva/enzymology , Larva/growth & development , Protein Kinase C-alpha
...